1. Academic Validation
  2. NMI inhibits cancer stem cell traits by downregulating hTERT in breast cancer

NMI inhibits cancer stem cell traits by downregulating hTERT in breast cancer

  • Cell Death Dis. 2017 May 11;8(5):e2783. doi: 10.1038/cddis.2017.200.
Xu Feng 1 2 Xiangdong Xu 3 Xiangsheng Xiao 2 Kun Zou 1 Wendan Yu 1 Jiali Wu 2 Ranran Tang 1 Yue Gao 1 Jiaojiao Hao 1 Xinrui Zhao 1 Yina Liao 1 Yiming Chen 1 Wenlin Huang 2 4 Wei Guo 1 Lan Kang 1 Wuguo Deng 2 4
Affiliations

Affiliations

  • 1 Institute of Cancer Stem Cell & The First Affiliated Hospital, Dalian Medical University, Dalian, China.
  • 2 Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China.
  • 3 Department of Thyroid & Breast Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
  • 4 State Key Laboratory of Targeted Drug for Tumors of Guangdong Province, Guangzhou Double Bioproduct Inc., Guangzhou, China.
Abstract

N-myc and STAT interactor (NMI) has been proved to bind to different transcription factors to regulate a variety of signaling mechanisms including DNA damage, cell cycle and epithelial-mesenchymal transition. However, the role of NMI in the regulation of Cancer Stem Cells (CSCs) remains poorly understood. In this study, we investigated the regulation of NMI on CSCs traits in breast Cancer and uncovered the underlying molecular mechanisms. We found that NMI was lowly expressed in breast Cancer Stem Cells (BCSCs)-enriched populations. Knockdown of NMI promoted CSCs traits while its overexpression inhibited CSCs traits, including the expression of CSC-related markers, the number of CD44+CD24- cell populations and the ability of mammospheres formation. We also found that NMI-mediated regulation of BCSCs traits was at least partially realized through the modulation of hTERT signaling. NMI knockdown upregulated hTERT expression while its overexpression downregulated hTERT in breast Cancer cells, and the changes in CSCs traits and cell invasion ability mediated by NMI were rescued by hTERT. The in vivo study also validated that NMI knockdown promoted breast Cancer growth by upregulating hTERT signaling in a mouse model. Moreover, further analyses for the clinical samples demonstrated that NMI expression was negatively correlated with hTERT expression and the low NMI/high hTERT expression was associated with the worse status of clinical TNM stages in breast Cancer patients. Furthermore, we demonstrated that the interaction of YY1 protein with NMI and its involvement in NMI-mediated transcriptional regulation of hTERT in breast Cancer cells. Collectively, our results provide new insights into understanding the regulatory mechanism of CSCs and suggest that the NMI-YY1-hTERT signaling axis may be a potential therapeutic target for breast cancers.

Figures
Products